AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (4.2 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Open Access

Trimethylamine N-oxide generation process was influenced by the proportion and source of macronutrients in the diet

Chengcheng Wanga,Xuefeng DuanaXiaoyue LiaJinyue YangaChanghu Xuea,bTeruyoshi YanagitacTiantian Zhanga( )Yuming Wanga,b( )
College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, China
Laboratory of Nutrition Biochemistry, Department of Applied Biochemistry and Food Science, Saga University, Saga 840-8502, Japan

Peer review under responsibility of Tsinghua University Press.

Show Author Information

Highlights

• TMAO generation was increased by low non-meat protein and high fat.

• TMAO generation was decreased by plant protein and refined sugar.

• Plant oil decreased TMA production but increased TMAO generation from TMA.

• Atherosclerosis might be influenced by dietary composition but not elevated TMAO.

Graphical Abstract

Abstract

Trimethylamine N-oxide (TMAO) is a risk factor of various chronic diseases, which was produced by metabolism from precursors to trimethylamine (TMA) in gut and the oxidation from TMA in liver. The TMA generation was influenced by diet, mainly due to the rich TMAO precursors in diet. However, it was still unclear that the effects of different proportion and source of macronutrients in different dietary pattern on the production process of TMAO. Here, the generation of TMA from precursors and TMAO from TMA was determined after single oral choline chloride and intraperitoneal injection TMA, respectively, in mice fed with carbohydrates, proteins and fats in different proportion and sources. The results suggested that the generation of TMAO was increased by low non-meat protein and high fat via enhancing the production of TMAO from TMA, and decreased by plant protein and refined sugar via reducing TMA production from precursors in gut and TMAO transformation from TMA in liver. The high fat and high sugar diets accelerating the development of atherosclerosis did not increase the production of TMAO, the risk factor for atherosclerosis, which indicated that the dietary compositions rather than the elevated TMAO level might be a more key risk factor for atherosclerosis.

References

[1]

Z. Wang, E. Klipfell, B.J. Bennett, et al., Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease, Nature 472 (2011) 57-63. http://doi.org/10.1038/nature09922.

[2]

R.A. Koeth, Z. Wang, B.S. Levison, et al., Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis, Nat. Med. 19 (2013) 576-585. http://doi.org/10.1038/nm.3145.

[3]

Z.L. Yu, L.Y. Zhang, X.M. Jiang, et al., Effects of dietary choline, betaine, and L-carnitine on the generation of trimethylamine-N-oxide in healthy mice, J. Food Sci. 85 (2020) 2207-2215. https://doi.org/10.1111/1750-3841.15186.

[4]

X.F. Duan, C.C. Wang, C.H. Xue, et al., Trimethylamine N-oxide generation from choline-containing precursors is closely associated with their molecular structure, J. Agric. Food Chem. 69 (2021) 2933-2935. http://doi.org/10.1021/acs.jafc.1c01068.

[5]

X. Gao, C. Jiang, J. Xu, et al., Serum pharmacokinetics of choline, trimethylamine, and trimethylamine-N-oxide after oral gavage of phosphatidylcholines with different fatty acid compositions in mice, Biosci. Biotechnol. Biochem. 80 (2016) 2217-2223. http://doi.org/10.1080/09168451.2016.1206813.

[6]

S. Krishnan, L.E. O’Connor, Y. Wang, et al., Adopting a Mediterranean-style eating pattern with low, but not moderate, unprocessed, lean red meat intake reduces fasting serum trimethylamine N-oxide (TMAO) in adults who are overweight or obese, Br. J. Nutr. 128 (2021) 1-21. http://doi.org/10.1017/s0007114521004694.

[7]

Z. Wang, N. Bergeron, B.S. Levison, et al., Impact of chronic dietary red meat, white meat, or non-meat protein on trimethylamine N-oxide metabolism and renal excretion in healthy men and women, Eur. Heart J. 40 (2019) 583-594. http://doi.org/10.1093/eurheartj/ehy799.

[8]

L. Ding, L.Y. Zhang, H.H. Shi, et al., Eicosapentaenoic acid-enriched phosphoethanolamine plasmalogens alleviated atherosclerosis by remodeling gut microbiota to regulate bile acid metabolism in LDLR–/– Mice, J. Agric. Food Chem. 68 (2020) 5339-5348. http://doi.org/10.1021/acs.jafc.9b08296.

[9]

L.A. Bolte, A. Vich Vila, F. Imhann, et al., Long-term dietary patterns are associated with pro-inflammatory and anti-inflammatory features of the gut microbiome, Gut 70 (2021) 1287-1298. http://doi.org/10.1136/gutjnl-2020-322670.

[10]

J. Yu, T. Zhang, X. Gao, et al., Fish oil affects the metabolic process of trimethylamine N-oxide precursor through trimethylamine production and flavin-containing monooxygenase activity in male C57BL/6 mice, RSC Adv. 7 (2017) 56655-56661. http://doi.org/10.1039/C7RA10248H.

[11]

D.M. Shih, Z. Wang, R. Lee, et al., Flavin containing monooxygenase 3 exerts broad effects on glucose and lipid metabolism and atherosclerosis, J. Lipid Res. 56 (2015) 22-37. http://doi.org/10.1194/jlr.M051680.

[12]

C.C. Wang, L. Du, H.H. Shi, et al., Dietary EPA-enriched phospholipids alleviate chronic stress and LPS-induced depression- and anxiety-like behavior by regulating immunity and neuroinflammation, Mol. Nutr. Food Res. 65 (2021) 2100009. http://doi.org/https://doi.org/10.1002/mnfr.202100009.

[13]

G. Falony, S. Vieira-Silva, J. Raes, Microbiology meets big data: the case of gut microbiota-derived trimethylamine, Annu. Rev. Microbiol. 69 (2015) 305-321. http://doi.org/10.1146/annurev-micro-091014-104422.

[14]

C.E. Cho, S. Taesuwan, O.V. Malysheva, et al., Trimethylamine-N-oxide (TMAO) response to animal source foods varies among healthy young men and is influenced by their gut microbiota composition: a randomized controlled trial, Mol. Nutr. Food Res. 61 (2017) 1600324. http://doi.org/10.1002/mnfr.201600324.

[15]

S. Rath, B. Heidrich, D.H. Pieper, et al., Uncovering the trimethylamine-producing bacteria of the human gut microbiota, Microbiome 5 (2017) 54. http://doi.org/10.1186/s40168-017-0271-9.

[16]

E.P. Treacy, B.R. Akerman, L.M. Chow, et al., Mutations of the flavin-containing monooxygenase gene (FMO3) cause trimethylaminuria, a defect in detoxication, Hum. Mol. Genet. 7 (1998) 839-845. http://doi.org/10.1093/hmg/7.5.839.

[17]

A. Cotillard, S.P. Kennedy, L.C. Kong, et al., Dietary intervention impact on gut microbial gene richness, Nature 500 (2013) 585-588. http://doi.org/10.1038/nature12480.

[18]

S.H. Zeisel, M.H. Mar, J.C. Howe, et al., Concentrations of choline-containing compounds and betaine in common foods, J. Nutr. 133 (2003) 1302-1307. http://doi.org/10.1093/jn/133.5.1302.

[19]

K. Chen, X. Zheng, M. Feng, et al., Gut microbiota-dependent metabolite trimethylamine N-oxide contributes to cardiac dysfunction in western diet-induced obese mice, Front. Physiol. 8 (2017) 139. http://doi.org/10.3389/fphys.2017.00139.

[20]

N.E. Boutagy, A.P. Neilson, K.L. Osterberg, et al., Short-term high-fat diet increases postprandial trimethylamine-N-oxide in humans, Nutr Res. 35 (2015) 858-864. http://doi.org/10.1016/j.nutres.2015.07.002.

[21]

Y. Wan, F. Wang, J. Yuan, et al., Effects of dietary fat on gut microbiota and faecal metabolites, and their relationship with cardiometabolic risk factors: a 6-month randomised controlled-feeding trial, Gut 68 (2019) 1417-1429. http://doi.org/10.1136/gutjnl-2018-317609.

[22]

N. Bergeron, P.T. Williams, R. Lamendella, et al., Diets high in resistant starch increase plasma levels of trimethylamine-N-oxide, a gut microbiome metabolite associated with CVD risk, Br. J. Nutr. 116 (2016) 2020-2029. http://doi.org/10.1017/s0007114516004165.

[23]

A. Genoni, C.T. Christophersen, J. Lo, et al., Long-term Paleolithic diet is associated with lower resistant starch intake, different gut microbiota composition and increased serum TMAO concentrations, Eur. J. Nutr. 59 (2020) 1845-1858. http://doi.org/10.1007/s00394-019-02036-y.

[24]

J.A. Kemp, H.F. dos Santos, H.E. de Jesus, et al., Resistant starch type-2 supplementation does not decrease trimethylamine N-oxide (TMAO) plasma level in hemodialysis patients, J. Am. Nutr. Assoc. 41 (2022) 1-8. http://doi.org/10.1080/07315724.2021.1967814.

[25]

A. Genoni, J. Lo, P. Lyons-Wall, et al., A Paleolithic diet lowers resistant starch intake but does not affect serum trimethylamine-N-oxide concentrations in healthy women, Br. J. Nutr. 121 (2019) 322-329. http://doi.org/10.1017/s000711451800329x.

[26]

S. Li, J. You, Z. Wang, et al., Curcumin alleviates high-fat diet-induced hepatic steatosis and obesity in association with modulation of gut microbiota in mice, Food Res. Int. 143 (2021) 110270. http://doi.org/10.1016/j.foodres.2021.110270.

[27]

B.J. Bennett, T.Q. de Aguiar Vallim, Z. Wang, et al., Trimethylamine-N-oxide, a metabolite associated with atherosclerosis, exhibits complex genetic and dietary regulation, Cell Metab. 17 (2013) 49-60. http://doi.org/10.1016/j.cmet.2012.12.011.

[28]

J.M. Ridlon, D.J. Kang, P.B. Hylemon, et al., Bile acids and the gut microbiome, Curr. Opin. Gastroenterol. 30 (2014) 332-338. http://doi.org/10.1097/mog.0000000000000057.

[29]

B.M. Liao, S.A. Mcmanus, W.E. Hughes, et al., Flavin-containing monooxygenase 3 reduces endoplasmic reticulum stress in lipid-treated hepatocytes, Mol. Endocrinol. 30 (2016) 417-428. http://doi.org/10.1210/me.2015-1217.

[30]

G.S. Hotamisligil, Endoplasmic reticulum stress and the inflammatory basis of metabolic disease, Cell 140 (2010) 900-917. http://doi.org/10.1016/j.cell.2010.02.034.

[31]

S. Fu, S.M. Watkins, G.S. Hotamisligil, The role of endoplasmic reticulum in hepatic lipid homeostasis and stress signaling, Cell Metab. 15 (2012) 623-634. http://doi.org/10.116/j.cmet.2012.03.007.

[32]

Y. Zhao, N. Yang, J. Gao, et al., The effect of different l-carnitine administration routes on the development of atherosclerosis in ApoE knockout mice, Mol. Nutr. Food Res. 62 (2018) 299. http://doi.org/10.1002/mnfr.201700299.

[33]

J.Y. Yang, T.T. Zhang, Z.L. Yu, et al., Taurine alleviates trimethylamine N-oxide-induced atherosclerosis by regulating bile acid metabolism in ApoE–/– mice, J. Agric. Food Chem. 70 (2022) 5738-5747. http://doi.org/10.1021/acs.jafc.2c01376.

[34]

L. Ding, M. Chang, Y. Guo, et al., Trimethylamine-N-oxide (TMAO)-induced atherosclerosis is associated with bile acid metabolism, Lipids Health Dis. 17 (2018) 286. http://doi.org/10.1186/s12944-018-0939-6.

[35]

P. Aldana-Hernández, K.A. Leonard, Y.Y. Zhao, et al., Dietary choline or trimethylamine N-oxide supplementation does not influence atherosclerosis development in Ldlr-/- and Apoe-/- male mice, J. Nutr. 150 (2019) 249-255. http://doi.org/10.1093/jn/nxz214.

[36]

Z.L. Yu, L. Ding, C.H. Xue, et al., Dietary trimethylamine N-oxide exacerbated atherosclerosis under a low-fat rather than high-fat diet, J. Agric. Food Chem. 68 (2020) 6789-6791. http://doi.org/10.1021/acs.jafc.0c03190.

[37]

S. He, F. Kahles, S. Rattik, et al., Gut intraepithelial T cells calibrate metabolism and accelerate cardiovascular disease, Nature 566 (2019) 115-119. http://doi.org/10.1038/s41586-018-0849-9.

[38]

X. Gao, X. Liu, J. Xu, et al., Dietary trimethylamine N-oxide exacerbates impaired glucose tolerance in mice fed a high fat diet, J. Biosci. Bioeng. 118 (2014) 476-481. http://doi.org/https://doi.org/10.1016/j.jbiosc.2014.03.001.

[39]

X. Gao, J. Xu, C. Jiang, et al., Fish oil ameliorates trimethylamine N-oxide-exacerbated glucose intolerance in high-fat diet-fed mice, Food Funct. 6 (2015) 1117-1125. http://doi.org/10.1039/C5FO00007F.

Food Science and Human Wellness
Pages 649-658
Cite this article:
Wang C, Duan X, Li X, et al. Trimethylamine N-oxide generation process was influenced by the proportion and source of macronutrients in the diet. Food Science and Human Wellness, 2024, 13(2): 649-658. https://doi.org/10.26599/FSHW.2022.9250054

1493

Views

204

Downloads

2

Crossref

1

Web of Science

1

Scopus

0

CSCD

Altmetrics

Received: 25 July 2022
Revised: 25 September 2022
Accepted: 16 October 2022
Published: 25 September 2023
© 2024 Beijing Academy of Food Sciences. Publishing services by Tsinghua University Press.

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return